Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Mol Genet Metab ; 143(1-2): 108562, 2024 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-39121793

RESUMEN

Alkaptonuria is a rare disorder of tyrosine catabolism caused by deficiency of homogentisate 1,2-dioxygenase that leads to accumulation of homogentisic acid (HGA). Deposition of HGA-derived polymers in connective tissue causes progressive arthropathy of the spine and large joints, cardiac valvular disease, and genitourinary stones beginning in the fourth decade of life. Nitisinone, a potent inhibitor of the upstream enzyme, 4-hydroxyphenylpyruvate dioxygenase, dramatically reduces HGA production. As such, nitisinone is a proposed treatment for alkaptonuria. A randomized clinical trial of nitisinone in alkaptonuria confirmed the biochemical efficacy and tolerability of nitisinone for patients with alkaptonuria but the selected primary outcome did not demonstrate significant clinical benefit. Given that alkaptonuria is a rare disease with slow progression and variable presentation, identifying outcome parameters that can detect significant change during a time-limited clinical trial is challenging. To gain insight into patient-perceived improvements in quality of life and corresponding changes in physical function associated with nitisinone use, we conducted a post-hoc per protocol analysis of patient-reported outcomes and a functional assessment. Analysis revealed that nitisinone-treated patients showed significant improvements in complementary domains of the 36-Item Short-Form Survey (SF-36) and 6-min walk test (6MWT). Together, these findings suggest that nitisinone improves both quality of life and function of patients with alkaptonuria. The observed trends support nitisinone as a therapy for alkaptonuria.

2.
Mult Scler Relat Disord ; 82: 105393, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38181697

RESUMEN

Our article "Fingolimod: Assay analysis of US generic capsule products reveals variation in fingolimod content beyond the recommended acceptance criteria" highlighted the variation of active ingredient in generic fingolimod capsule products. This analysis was prompted by reports of clinical adverse events and/or multiple sclerosis relapse in patients following transition from Gilenya® fingolimod capsules (Novartis) to generic fingolimod capsule products. Further assay analysis functioned to both confirm previous out-of-specification findings, and to identify an additional generic product that failed to comply with United States Pharmacopeia (USP) recommendations.


Asunto(s)
Esclerosis Múltiple Recurrente-Remitente , Esclerosis Múltiple , Humanos , Clorhidrato de Fingolimod/uso terapéutico , Inmunosupresores/uso terapéutico , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/inducido químicamente , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inducido químicamente
3.
Nat Metab ; 5(11): 1870-1886, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37946084

RESUMEN

Tumors are intrinsically heterogeneous and it is well established that this directs their evolution, hinders their classification and frustrates therapy1-3. Consequently, spatially resolved omics-level analyses are gaining traction4-9. Despite considerable therapeutic interest, tumor metabolism has been lagging behind this development and there is a paucity of data regarding its spatial organization. To address this shortcoming, we set out to study the local metabolic effects of the oncogene c-MYC, a pleiotropic transcription factor that accumulates with tumor progression and influences metabolism10,11. Through correlative mass spectrometry imaging, we show that pantothenic acid (vitamin B5) associates with MYC-high areas within both human and murine mammary tumors, where its conversion to coenzyme A fuels Krebs cycle activity. Mechanistically, we show that this is accomplished by MYC-mediated upregulation of its multivitamin transporter SLC5A6. Notably, we show that SLC5A6 over-expression alone can induce increased cell growth and a shift toward biosynthesis, whereas conversely, dietary restriction of pantothenic acid leads to a reversal of many MYC-mediated metabolic changes and results in hampered tumor growth. Our work thus establishes the availability of vitamins and cofactors as a potential bottleneck in tumor progression, which can be exploited therapeutically. Overall, we show that a spatial understanding of local metabolism facilitates the identification of clinically relevant, tractable metabolic targets.


Asunto(s)
Neoplasias de la Mama , Humanos , Ratones , Animales , Femenino , Neoplasias de la Mama/metabolismo , Ácido Pantoténico , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción/metabolismo , Vitaminas
4.
Mult Scler Relat Disord ; 80: 105077, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37864877

RESUMEN

The immunomodulating agent fingolimod is a sphingosine-1-phosphate receptor modulator used in the treatment of multiple sclerosis (MS). We analyzed three FDA approved fingolimod 0.5 mg generic capsule products for fingolimod content. Assay results demonstrated a wide variation in fingolimod content between manufacturers, with one product demonstrating a fingolimod content of 76.8 % of the approved dose. This falls significantly below the FDA acceptance criteria of 90.0-110.0 % of label claim.


Asunto(s)
Clorhidrato de Fingolimod , Esclerosis Múltiple , Humanos , Clorhidrato de Fingolimod/uso terapéutico , Inmunosupresores/uso terapéutico , Esfingosina , Esclerosis Múltiple/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico
5.
Clin Nurse Spec ; 37(4): 177-186, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37410562

RESUMEN

PURPOSE: The purpose of this study was to examine the perceived self-efficacy of the clinical nurse specialist working in the United States during the COVID-19 pandemic and explore whether there was any difference in self-efficacy based on practice focus (spheres of impact) and if differences existed between self-efficacy and demographics. DESIGN: This study used a nonexperimental, correlational, cross-sectional design utilizing a voluntary, anonymous, 1-time survey administered through Qualtrics (Qualtrics, Provo, UT). METHODS: The National Association of Clinical Nurse Specialists and 9 state affiliates distributed the electronic survey starting late October 2021 through January 2022. Survey content consisted of demographics and the General Self-efficacy Scale, which measures the individual's perceived ability to cope and execute tasks when faced with hardship or adversity. Sample size was 105. RESULTS: Results included a high perception of self-efficacy of the clinical nurse specialist working during the pandemic, no statistical significance in practice focus, and a statistically significant difference in the scores of self-efficacy for participants with previous infectious disease experience compared with those without experience. CONCLUSIONS: Clinical nurse specialists with previous infectious disease experience can guide policy, be utilized in multifaceted roles to support future infectious disease outbreaks, and develop training to prepare and support clinicians during crises such as pandemics.


Asunto(s)
COVID-19 , Enfermeras Clínicas , Humanos , Estados Unidos/epidemiología , COVID-19/epidemiología , Pandemias , Autoeficacia , Estudios Transversales , Encuestas y Cuestionarios
6.
Nat Commun ; 12(1): 1998, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33790302

RESUMEN

The heterogeneity of breast cancer plays a major role in drug response and resistance and has been extensively characterized at the genomic level. Here, a single-cell breast cancer mass cytometry (BCMC) panel is optimized to identify cell phenotypes and their oncogenic signalling states in a biobank of patient-derived tumour xenograft (PDTX) models representing the diversity of human breast cancer. The BCMC panel identifies 13 cellular phenotypes (11 human and 2 murine), associated with both breast cancer subtypes and specific genomic features. Pre-treatment cellular phenotypic composition is a determinant of response to anticancer therapies. Single-cell profiling also reveals drug-induced cellular phenotypic dynamics, unravelling previously unnoticed intra-tumour response diversity. The comprehensive view of the landscapes of cellular phenotypic heterogeneity in PDTXs uncovered by the BCMC panel, which is mirrored in primary human tumours, has profound implications for understanding and predicting therapy response and resistance.


Asunto(s)
Benzamidas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Xenoinjertos/efectos de los fármacos , Morfolinas/farmacología , Piperazinas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Xenoinjertos/metabolismo , Humanos , Células MCF-7 , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Inhibidores de Proteínas Quinasas/farmacología , Resultado del Tratamiento
7.
BMC Genomics ; 19(1): 19, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29304755

RESUMEN

BACKGROUND: Patient-Derived Tumour Xenografts (PDTXs) have emerged as the pre-clinical models that best represent clinical tumour diversity and intra-tumour heterogeneity. The molecular characterization of PDTXs using High-Throughput Sequencing (HTS) is essential; however, the presence of mouse stroma is challenging for HTS data analysis. Indeed, the high homology between the two genomes results in a proportion of mouse reads being mapped as human. RESULTS: In this study we generated Whole Exome Sequencing (WES), Reduced Representation Bisulfite Sequencing (RRBS) and RNA sequencing (RNA-seq) data from samples with known mixtures of mouse and human DNA or RNA and from a cohort of human breast cancers and their derived PDTXs. We show that using an In silico Combined human-mouse Reference Genome (ICRG) for alignment discriminates between human and mouse reads with up to 99.9% accuracy and decreases the number of false positive somatic mutations caused by misalignment by >99.9%. We also derived a model to estimate the human DNA content in independent PDTX samples. For RNA-seq and RRBS data analysis, the use of the ICRG allows dissecting computationally the transcriptome and methylome of human tumour cells and mouse stroma. In a direct comparison with previously reported approaches, our method showed similar or higher accuracy while requiring significantly less computing time. CONCLUSIONS: The computational pipeline we describe here is a valuable tool for the molecular analysis of PDTXs as well as any other mixture of DNA or RNA species.


Asunto(s)
Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Perfilación de la Expresión Génica , Humanos , Ratones , Mutación , Alineación de Secuencia , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN
8.
Endocr Relat Cancer ; 23(12): T259-T270, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27702751

RESUMEN

Despite remarkable advances in our understanding of the drivers of human malignancies, new targeted therapies often fail to show sufficient efficacy in clinical trials. Indeed, the cost of bringing a new agent to market has risen substantially in the last several decades, in part fuelled by extensive reliance on preclinical models that fail to accurately reflect tumour heterogeneity. To halt unsustainable rates of attrition in the drug discovery process, we must develop a new generation of preclinical models capable of reflecting the heterogeneity of varying degrees of complexity found in human cancers. Patient-derived tumour xenograft (PDTX) models prevail as arguably the most powerful in this regard because they capture cancer's heterogeneous nature. Herein, we review current breast cancer models and their use in the drug discovery process, before discussing best practices for developing a highly annotated cohort of PDTX models. We describe the importance of extensive multidimensional molecular and functional characterisation of models and combination drug-drug screens to identify complex biomarkers of drug resistance and response. We reflect on our own experiences and propose the use of a cost-effective intermediate pharmacogenomic platform (the PDTX-PDTC platform) for breast cancer drug and biomarker discovery. We discuss the limitations and unanswered questions of PDTX models; yet, still strongly envision that their use in basic and translational research will dramatically change our understanding of breast cancer biology and how to more effectively treat it.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Medicina de Precisión/métodos , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Neoplasias de la Mama/patología , Femenino , Humanos , Neoplasias Mamarias Experimentales/patología , Trasplante de Neoplasias/métodos
9.
Cell ; 167(1): 260-274.e22, 2016 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-27641504

RESUMEN

The inter- and intra-tumor heterogeneity of breast cancer needs to be adequately captured in pre-clinical models. We have created a large collection of breast cancer patient-derived tumor xenografts (PDTXs), in which the morphological and molecular characteristics of the originating tumor are preserved through passaging in the mouse. An integrated platform combining in vivo maintenance of these PDTXs along with short-term cultures of PDTX-derived tumor cells (PDTCs) was optimized. Remarkably, the intra-tumor genomic clonal architecture present in the originating breast cancers was mostly preserved upon serial passaging in xenografts and in short-term cultured PDTCs. We assessed drug responses in PDTCs on a high-throughput platform and validated several ex vivo responses in vivo. The biobank represents a powerful resource for pre-clinical breast cancer pharmacogenomic studies (http://caldaslab.cruk.cam.ac.uk/bcape), including identification of biomarkers of response or resistance.


Asunto(s)
Bancos de Muestras Biológicas , Neoplasias de la Mama , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Biomarcadores Farmacológicos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/genética , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Pruebas de Farmacogenómica , Células Tumorales Cultivadas
10.
Cell Rep ; 13(11): 2480-2490, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26686634

RESUMEN

The transforming growth factor beta (TGF-ß) signaling pathway exerts opposing effects on cancer cells, acting as either a tumor promoter or a tumor suppressor. Here, we show that these opposing effects are a result of the synergy between SMAD3, a downstream effector of TGF-ß signaling, and the distinct epigenomes of breast-tumor-initiating cells (BTICs). These effects of TGF-ß are associated with distinct gene expression programs, but genomic SMAD3 binding patterns are highly similar in the BTIC-promoting and BTIC-suppressing contexts. Our data show cell-type-specific patterns of DNA and histone modifications provide a modulatory layer by determining accessibility of genes to regulation by TGF-ß/SMAD3. LBH, one such context-specific target gene, is regulated according to its DNA methylation status and is crucial for TGF-ß-dependent promotion of BTICs. Overall, these results reveal that the epigenome plays a central and previously overlooked role in shaping the context-specific effects of TGF-ß in cancer.


Asunto(s)
Transducción de Señal/efectos de los fármacos , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Sitios de Unión , Línea Celular Tumoral , ADN/química , ADN/metabolismo , Metilación de ADN , Epigénesis Genética , Histonas/metabolismo , Humanos , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína Smad2/metabolismo , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
11.
Nature ; 518(7539): 422-6, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25470049

RESUMEN

Human cancers, including breast cancers, comprise clones differing in mutation content. Clones evolve dynamically in space and time following principles of Darwinian evolution, underpinning important emergent features such as drug resistance and metastasis. Human breast cancer xenoengraftment is used as a means of capturing and studying tumour biology, and breast tumour xenografts are generally assumed to be reasonable models of the originating tumours. However, the consequences and reproducibility of engraftment and propagation on the genomic clonal architecture of tumours have not been systematically examined at single-cell resolution. Here we show, using deep-genome and single-cell sequencing methods, the clonal dynamics of initial engraftment and subsequent serial propagation of primary and metastatic human breast cancers in immunodeficient mice. In all 15 cases examined, clonal selection on engraftment was observed in both primary and metastatic breast tumours, varying in degree from extreme selective engraftment of minor (<5% of starting population) clones to moderate, polyclonal engraftment. Furthermore, ongoing clonal dynamics during serial passaging is a feature of tumours experiencing modest initial selection. Through single-cell sequencing, we show that major mutation clusters estimated from tumour population sequencing relate predictably to the most abundant clonal genotypes, even in clonally complex and rapidly evolving cases. Finally, we show that similar clonal expansion patterns can emerge in independent grafts of the same starting tumour population, indicating that genomic aberrations can be reproducible determinants of evolutionary trajectories. Our results show that measurement of genomically defined clonal population dynamics will be highly informative for functional studies using patient-derived breast cancer xenoengraftment.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Células Clonales/metabolismo , Células Clonales/patología , Genoma Humano/genética , Análisis de la Célula Individual , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Neoplasias de la Mama/secundario , Análisis Mutacional de ADN , Genómica , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Trasplante de Neoplasias , Factores de Tiempo , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
12.
Nat Commun ; 3: 1055, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22968701

RESUMEN

The role of transforming growth factor-beta (TGFß) in the progression of different molecular subtypes of breast cancer has not been clarified. Here we show that TGFß increases breast tumour-initiating cell (BTIC) numbers but only in claudin(low) breast cancer cell lines by orchestrating a specific gene signature enriched in stem cell processes that predicts worse clinical outcome in breast cancer patients. NEDD9, a member of the Cas family of integrin scaffold proteins, is necessary to mediate these TGFß-specific effects through a positive feedback loop that integrates TGFß/Smad and Rho-actin-SRF-dependent signals. In normal human mammary epithelium, TGFß induces progenitor activity only in the basal/stem cell compartment, where claudin(low) cancers are presumed to arise. These data show opposing responses to TGFß in both breast malignant cell subtypes and normal mammary epithelial cell subpopulations and suggest therapeutic strategies for a subset of human breast cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Claudinas/metabolismo , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Inmunoprecipitación de Cromatina , Claudinas/genética , Femenino , Humanos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Tumorales Cultivadas
13.
EMBO Mol Med ; 3(3): 167-80, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21337521

RESUMEN

The telomeric amplicon at 8p12 is common in oestrogen receptor-positive (ER+) breast cancers. Array-CGH and expression analyses of 1172 primary breast tumours revealed that ZNF703 was the single gene within the minimal amplicon and was amplified predominantly in the Luminal B subtype. Amplification was shown to correlate with increased gene and protein expression and was associated with a distinct expression signature and poor clinical outcome. ZNF703 transformed NIH 3T3 fibroblasts, behaving as a classical oncogene, and regulated proliferation in human luminal breast cancer cell lines and immortalized human mammary epithelial cells. Manipulation of ZNF703 expression in the luminal MCF7 cell line modified the effects of TGFß on proliferation. Overexpression of ZNF703 in normal human breast epithelial cells enhanced the frequency of in vitro colony-forming cells from luminal progenitors. Taken together, these data strongly point to ZNF703 as a novel oncogene in Luminal B breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas Portadoras/metabolismo , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/metabolismo , Proteínas Oncogénicas/metabolismo , Animales , Línea Celular , Proliferación Celular , Células Epiteliales , Femenino , Fibroblastos , Perfilación de la Expresión Génica , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA